Modulation of epileptogenesis: A paradigm for the integration of enzyme-based microelectrode arrays and optogenetics

Modulation of epileptogenesis: A paradigm for the integration of enzyme-based microelectrode arrays and optogenetics

Journal Pre-proof Modulation of epileptogenesis: A paradigm for the integration of enzyme-based microelectrode arrays and optogenetics Corwin R. Butle...

3MB Sizes 0 Downloads 14 Views

Journal Pre-proof Modulation of epileptogenesis: A paradigm for the integration of enzyme-based microelectrode arrays and optogenetics Corwin R. Butler, Jeffery A. Boychuk, Francois Pomerleau, R. Alcala, P. Huettl, Y. Ai, J. Jakobsson, Sidney W. Whiteheart, Greg A. Gerhardt, Bret N. Smith, John T. Slevin

PII:

S0920-1211(19)30507-8

DOI:

https://doi.org/10.1016/j.eplepsyres.2019.106244

Reference:

EPIRES 106244

To appear in:

Epilepsy Research

Received Date:

21 September 2019

Revised Date:

22 October 2019

Accepted Date:

22 November 2019

Please cite this article as: Butler CR, Boychuk JA, Pomerleau F, Alcala R, Huettl P, Ai Y, Jakobsson J, Whiteheart SW, Gerhardt GA, Smith BN, Slevin JT, Modulation of epileptogenesis: A paradigm for the integration of enzyme-based microelectrode arrays and optogenetics, Epilepsy Research (2019), doi: https://doi.org/10.1016/j.eplepsyres.2019.106244

This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. © 2019 Published by Elsevier.

Modulation of epileptogenesis: A paradigm for the integration of enzyme-based microelectrode arrays and optogenetics Corwin R. Butlera*, Jeffery A. Boychuka,b*, Francois Pomerleaud,h, R. Alcalae, P. Huettld,h, Y. Aid, J. Jakobssoni, Sidney W. Whiteheartf,g, Greg A. Gerhardtb,c,d,h, Bret N. Smitha,b,c,d, & John T. Slevinb,e,g,h

re

-p

ro

of

a. Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536 b. Epilepsy Center, University of Kentucky, Lexington, KY 40536 c. Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY 40536 d. Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY 40536 e. Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY 40536 f. Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536 g. Veterans Affairs Medical Center, Lexington, KY 40536 h. Brain Restoration Center, University of Kentucky, Lexington, KY 40356 i. Wallenburg Neuroscience Center, Lund Stem Cell Center, Lund University, Lund, Sweden * Authors contributed equally

ur na

lP

Corresponding author: John T. Slevin Kentucky Clinic J-401 University of Kentucky College of Medicine 740 South Limestone Street Lexington, KY 40536-0284 [email protected] Number of words in abstract:

250

Jo

Contents of Supplemental Material: none

1

Highlights: 

Distribution of transfected opsins occurred bilaterally, with contralateral expression appearing mainly in the commissural projections of mossy cells and pyramidal neurons.



Transfection of ChR2 primarily occurred in glutamatergic neurons, while transfection of NpHR resulted in GABAergic neuron transfection. Activation of ChR2 resulted in frequency- and duration-dependent release of glutamate.



Use of viral vectors to deliver channelrhodopsin and halorhodopsin to dentate gyrus

of





ro

provided excitatory and inhibitory control of dentate gyrus granule cells.

Light stimulation of dentate gyrus in ChR2-transfected rats was associated with

-p

epileptogenesis; light stimulation of dentate gyrus in NpHR-transfected rats retarded

re

classical amygdalar kindling.

lP

Abstract

Background: Genesis of acquired epilepsy includes transformations spanning genetic-to-

ur na

network-level modifications, disrupting the regional excitatory/inhibitory balance. Methodology concurrently tracking changes at multiple levels is lacking. Here, viral vectors are used to differentially express two opsin proteins in neuronal populations within dentate gyrus (DG) of hippocampus. When activated, these opsins induced excitatory or inhibitory neural output that

Jo

differentially affected neural networks and epileptogenesis. In vivo measures included behavioral observation coupled to real-time measures of regional glutamate flux using ceramic-based amperometric microelectrode arrays (MEAs). Results: Using MEA technology, phasic increases of extracellular glutamate were recorded immediately upon application of blue light/488 nm to DG of rats previously transfected

2

with an AAV 2/5 vector containing an (excitatory) channelrhodopsin-2 transcript. Rats receiving twice-daily 30-sec light stimulation to DG ipsilateral to viral transfection progressed through Racine seizure stages. AAV 2/5 (inhibitory) halorhodopsin-transfected rats receiving concomitant amygdalar kindling and DG light stimuli were kindled significantly more slowly than non-stimulated controls. In in vitro slice preparations, both excitatory and inhibitory responses were independently evoked in dentate granule cells during appropriate light

of

stimulation. Latency to response and sensitivity of responses suggest a degree of neuron subtype-

ro

selective functional expression of the transcripts.

Conclusions: This study demonstrates the potential for coupling MEA technology and

-p

optogenetics for real-time neurotransmitter release measures and modification of seizure

re

susceptibility in animal models of epileptogenesis. This microelectrode/optogenetic technology could prove useful for characterization of network and system level dysfunction in diseases

ur na

of future treatment strategies.

lP

involving imbalanced excitatory/inhibitory control of neuron populations and guide development

Key Words

Jo

Dentate gyrus, epileptogenesis, glutamate, microelectrode array, optogenetics

3

1. Introduction

The precise mechanisms that drive epileptogenesis are unknown. Clearly, there are many potential triggering events. Changes occur over time at many levels of organization, including genetic alterations, molecular and biochemical modifications, alterations of channels and neurotransmission, morphologic and cellular reorganization, and network transformations. 1

of

Which of these processes is transcendent and whether any singular modification represents a

ro

common node for the multiple causations ultimately leading to the behavioral expression of

spontaneous seizures is unknown. Regardless, acquired epilepsy, the product of the process of

-p

epileptogenesis, is associated with increased likelihood of pathological hyper-synchronous

re

activation of large populations of neurons that may occur through imbalanced secretion of both excitatory (glutamate) and inhibitory (GABA) neurotransmitters. Understanding the contribution

lP

of cellular excitability to the process of epileptogenesis will be critical for developing diseasemodifying therapies to prevent the development of acquired epilepsy.

ur na

Specific, targeted control of neural systems to establish causality between neuronal activity and behavior has remained difficult to achieve until recent technological advances, including optogenetics, which introduces light sensitive proteins (opsins) into neurons that regulate transmembrane ion conductance 2, 3. The most common opsins in use modulate

Jo

membrane potentials, including blue light (488 nm)-activated channelrhodopsin (ChR2, excitation) and yellow light (593 nm)-activated halorhodopsin (NpHR, hyperpolarization) 4, 5. Most studies to date have reported the ability of opsin transfection to modify behavior and ex vivo synaptic function and some have investigated targeting of different opsin promoters to differing neuron subpopulations 6. Here, amperometric glutamate microelectrode recordings,

4

immunolabeling, whole cell patch-clamp electrophysiology, and behavioral responses to electrical kindling were used to investigate targeting of different opsins to glutamatergic and GABAergic neuronal populations within the hippocampus, using a single adenoviral serotype. This multipronged, temporal approach allows network, neuronal, and presynaptic release changes to be linked to biochemical, anatomical, electrophysiological, and behavioral outcomes associated with epileptogenesis. The functional and behavioral outcomes of transfecting these

ro

of

opsins is discussed.

2. Materials and Methods

-p

2.1 Viral construct and hippocampal viral injections: The viral vectors used in this study were

re

pseudotyped AAV2/5 vectors, where the transgene of interest is flanked by inverted terminal repeats of the AAV2 packaged in an AAV5 capsid. The AAV vectors were produced using a

lP

double-transfection method with the plasmid (pAAV-hSyn-hChR2(H134R)-EYFP or pAAVhSyn-NpHR3.0-EYFP) and the appropriate helper plasmid containing the essential adenoviral

ur na

packaging genes, as previously described 7. The final titers of the injected AAV vector suspensions were between 2.4e+14 and 2.8e+14 genome copies/mL. All animal procedures were approved by the University of Kentucky Institutional Animal Care and Use Committee. Male Sprague-Dawley rats (Harlan Laboratories, Indianapolis, IN,

Jo

U.S.A.) were used for these experiments. At 18-24 weeks of age, rats were anesthetized with isoflurane (1-3%) and placed in a stereotaxic frame. A midline incision was made on the scalp to expose the skull. A burr hole was made above the dentate gyrus (DG) of the hippocampus (from bregma: AP: -4.4 mm; ML: -2.2 mm; 8). A Hamilton syringe was loaded with 1 µL of the channelrhodopsin (488 nm; blue light activation; pAAV-hSyn-hChR2(H134R)-EYFP) or

5

halorhodopsin (593 nm; yellow light activation; pAAV-hSyn-NpHR3.0-EYFP) suspension (in PBS) and was lowered to the DG (DV: -4.0) where the suspension was slowly injected over 4 min. The tissue was allowed to rest for 5 min before the needle was slowly retracted. Five to 7 weeks was allowed before experimentation was started to allow for near maximal expression of the vector to occur 9.

of

2.2 In vivo amperometric measures of glutamate using microelectrode assemblies (MEAs):

ro

Amperometric recordings, (100 kHz data acquisition displayed at a display frequency of 10 Hz), were performed using the FAST16mkIII recording system (Fast Analytical Sensing Technology,

-p

Quanteon, LLC, Nicholasville, KY). Ceramic-based Microelectrode arrays (MEAs) consisting of

re

four platinum (Pt) recording sites measuring 15×333 μm arranged vertically in dual pairs (S2 conformation) were configured to measure glutamate as previously described 10. Briefly, a

lP

solution of 1 % Glutamate-oxidase (GluOx; US Biological, Salem, MA), 1 % bovine serum albumin (BSA; Sigma-Aldrich, St. Louis, MO) and 0.125 % glutaraldehyde (Glut; Sigma-

ur na

Aldrich), was prepared and coated onto the bottom pair of recording sites to allow for the conversion of glutamate to α-ketoglutarate and the reporter molecule, H2O2, which can be oxidized at the surface of the Pt with an applied potential of +0.7 V vs Ag/AgCl. The top pair of recording sites, or the sentinel sites, only received a coating of the BSA/Glut matrix. This coating

Jo

configuration allows for background measures and subtraction to obtain a self-referenced glutamate signal for measures of both tonic and phasic glutamate 10, 11. To make the electrode even more specific for glutamate and to eliminate electroactive interfering molecules such as ascorbic acid and dopamine from reaching the Pt sites, 1,3-phenylenediamine (mPD), a molecular size exclusion layer, was electroplated onto all 4 Pt recording sites 10. Before in vivo

6

use, an in vitro calibration was used to determine selectivity (glutamate vs. ascorbic acid), sensitivity (slope, in nAmps/µM), and limit of detection (in µM based on a signal-to-noise ratio of 3), as previously described 10.

2.3 MEA and optical fiber configuration: For this study, the transfected channelrhodopsin (ChR2) was stimulated locally by affixing a guide cannula (Plastics One Roanoke VA) to the

of

MEA. A fiber optic cannula (Ø200 µm Core, 0.39 NA; Thorlabs, Newton, NJ) was then inserted

ro

through the guide with the tip of the fiber positioned ~ 200 µm from the surface of the MEA in between all 4 Pt sites (see Figure 1). The laser source (OBIS LS 488 nm, 100mW, Coherent,

-p

Santa Clara, CA) was controlled through its analog port using a Master-8 pulse generator

re

(A.M.P.I., Jerusalem, Israel). Pulse trains (100 pulses, 40 Hz) with variable pulse durations (1-20 ms) generating light intensities between 1 to 20 mW or constant light stimulation of variable

lP

intensities (1-50 mW) was delivered locally through the fiber optic cannula.

ur na

2.4 Electrical kindling: Fourteen-week-old male Sprague-Dawley rats (Harlan Laboratories) had stimulating electrodes surgically implanted in the right amygdala [from bregma: AP, 2.8; ML, +4.8; DV , 8.5; nose bar, 3.3 (flat skull)] as previously described 12. Animals were each tested for their after discharge (AD) threshold prior to kindling. An S88 dual channel stimulator (Grass

Jo

Technologies, West Warwick, RI, U.S.A.) delivered a 1 sec train of biphasic square wave pulses (1 ms pulse duration, 60 Hz) starting at an initial 100 μA until an AD was recorded on electroencephalography (EEG); this AD threshold was used for subsequent stimulation. About one third of animals exhibited no behavioral activity, and two-thirds experienced facial clonus (stage 1) or head nodding (stage 2) at their established AD threshold. Animals were housed

7

individually in an enriched environment and maintained on a 12 h light/dark cycle with food and water available ad libitum. For amygdala kindling, rats that had received an injection of NpHR were also affixed with a waveguide cannula (Ø200 µm Core, 0.39 NA; Thorlabs, Newton, NJ) targeted in the DG and received a light stimulation (~ 2-3 sec duration; 593nm; ~50 mW; LaserGlow technologies) immediately before the start of the kindling stimulus (1 sec).

of

ChR2 “kindling”: Some ChR2 injected animals were implanted with a waveguide cannula

ro

(Ø200 µm Core, 0.39 NA; Thorlabs, Newton, NJ) targeted to the DG. Animals were stimulated (30 sec) with a blue light laser at 50 mW power (OBIS LS 488 nm, Coherent, Santa Clara, CA).

-p

Unless specifically stated, all animals were stimulated twice daily, 5 days per week, until they

re

experienced two consecutive Racine stage 5 seizures (tonic–clonic activity with loss of postural

lP

control/falling13).

2.5 Slice preparation: Slices used for electrophysiological studies were obtained from rats 6

ur na

weeks after viral inoculation (i.e., 24-30 weeks of age). Rats were deeply anesthetized by isoflurane inhalation (5%) and decapitated while anesthetized. The brain was removed and placed in ice-cold (2-4oC) oxygenated artificial cerebrospinal fluid (ACSF) containing, in mM: 124 NaCl, 3 KCl, 2 CaCl2, 26 NaHCO3, 1.3 MgCl2, 11 glucose and 1.4 NaH2PO4 equilibrated

Jo

with bubbling 95% O2-5% CO2 (pH 7.2-7.4). Brains were blocked and glued to a sectioning stage, and 350 μm-thick slices were cut in the coronal plane in cold, oxygenated ACSF using a vibrating microtome (Vibratome Series 1000; Technical Products International, St. Louis, MO). Slices were transferred to a chamber containing oxygenated ACSF at 32-34 oC, where they were

8

equilibrated for at least one hour prior to recording. Slices of the septal hippocampus were used in these experiments from viral injected rats and comparable slices from saline injected rats.

2.6 Whole cell recordings: Coronal hippocampal slices containing the dorsal third of the dentate gyrus were transferred to a recording chamber on an upright, fixed-stage microscope equipped with infrared, differential interference contrast optics (i.e., IR-DIC; Olympus BX50WI), where

of

they were perfused with continuously warmed (32-34oC) ACSF. Recordings were performed

ro

from dentate granule cells as described previously 14-17. Recording pipettes were pulled from borosilicate glass (1.65 mm outer diameter, 0.45 mm inner diameter; King Precision Glass,

-p

Claremont, CA) with a P-87 puller (Sutter Instrument, Novato, CA). The intracellular solution

re

contained (in mM): 130 K+-gluconate, 1 NaCl, 5 EGTA, 10 HEPES, 1 MgCl2, 1 CaCl2, 3 KOH, and 2 ATP. Open tip series resistance was 2-5 MOhms. Recordings were obtained using an

lP

Axon 700B amplifier (Molecular Devices), low-pass filtered at 6 kHz, digitized at 20 kHz with a Digidata 1550A (Molecular Devices), and acquired and analyzed using pClamp 10.5 programs

ur na

(Clampfit, Molecular Devices). Cells were voltage-clamped at -65mV for recordings of excitatory postsynaptic currents (EPSCs; ChR2=10 cells from 5 rats, NpHR=8 cells from 5 rats) or 0 mV for recordings of inhibitory postsynaptic currents (IPSCs; ChR2=5 cells from 3 rats) for 5-10 min to allow equilibration of pipette and intercellular solutions prior to data collection.

Jo

Recordings were made from one cell in a slice, so cell and slice numbers are equivalent. Optical stimulation of the dentate gyrus was performed using 488 nm light for ChR2 stimulation and 593 nm light for NpHR stimulation. Three stimulation durations were used to test activation of the opsins (16 ms, 50 ms, and 500 ms). Following recording of responses to optical activation, either tetrodotoxin (TTX; 2 μM; Tocris Bioscience; Minneapolis, MN) to block action potentials,

9

kynurenic acid (1 mM; Sigma Aldrich; St. Louis, MO) to block EPSCs, or bicuculline methiodide (30 μM; Tocris Bioscience; Minneapolis, MN) to block IPSCs was added to the circulating ACSF.

2.7 Analysis of 7SComplex: 7S Complex was measured from synaptosomal extracts as

of

previously described in detail 12, 18-21.

ro

2.8 Statistical Analysis:

2.8.1 Electrophysiology data analysis was performed using Graphpad Prism software (Graphpad

-p

Software; La Jolla, CA), using one-way ANOVA followed by Tukey’s post hoc tests. Unless

re

otherwise indicated, data are expressed as mean ± SEM, and significance was set at p<0.05.

lP

2.8.2 Microelectrode array analysis: Raw amperometric glutamate release data and event markers were analyzed using custom exported MatLab-based software, to determine maximum

ur na

amplitude, exponential rate of signal decay (k-1), uptake rate (k-1*amp, glutamate only), peak or trough area, rise/drop time (Trise/drop), and time for 80% of signal decay (T80), as we have previously described 22-24. Grouped results of single comparisons of these metrics were tested for normality with the Shapiro-Wilk normality test and analyzed using repeated measures

Jo

analysis of variance (ANOVA with Bonferroni post-hoc) or a two-tailed Student’s t-test when data were normally distributed. The Wilcoxon signed rank test for paired samples or the MannWhitney U test for unpaired samples was used when normally-distributed population responses could not be established.

10

3. Results

3.1 Distribution of opsins: Viral vector injections vary as to distribution of the targeted expression from the injection site. In this study we found that unilateral injection of ChR2 and NpHR not only resulted in expression within the ipsilateral dentate gyrus neurons but also spread to the contralateral hemisphere, likely from the commissural projections associated with

of

hippocampal mossy cells and pyramidal neurons. Transfection of ChR2 resulted in widespread

ro

labeling of both principal cells and interneurons (Figure 2). In addition, transfection of NpHR in the rat dentate gyrus resulted in co-immunolabeling primarily with GABAergic neurons,

-p

consistent with electrophysiological results (Figure 2, also see Figures 7, 8). We have evaluated

re

the durability and longevity of both ChR2 and NpHR proteins. Electrophysiological responses to light stimulation in hippocampal slices (Figures 4-6) remained robust for up to 15 weeks

lP

following viral inoculation, supported by continued demonstration of the GFP reporter protein

ur na

out to 15 weeks. This range encompassed the duration of the experiments.

3.2 Light-induced glutamate release: Action potential generation in glutamatergic neurons results in the release of glutamate in the synaptic cleft between the pre- and post-synaptic neurons and spillover into the extracellular space, where it is largely inactivated by low

Jo

affinity/high capacity uptake into glia.25 In theory, when a large population of neurons fire synchronized action potentials, a large release of glutamate would be detected in the terminal region/extracellular space of that neuron population, but to date, very few studies have examined neurotransmitter release combined with optical stimulation. In this study we attached an optical fiber (200 µm o.d. ~200 µm from the recording sites) to our ceramic-based Pt MEA, which was

11

configured to directly record tonic and phasic glutamate release 26, and lowered the assembly into the DG. Figure 3 shows precise optical control of glutamate release using variable light intensities (0.2 mW to 50 mW). There was clear indication that the amplitude of the glutamate release was dependently related to power and the duration of the light emission. Repeated stimulation at 1 minute intervals were extremely reproducible and did not show signs of tachyphylaxis.

of

Furthermore, at equivalent power delivered at the tip of the fiber optic cannula (~4 to 5 mW), we

ro

did not observe significant differences in the amount of glutamate release whether using a light pulse train (20 ms, 40 Hz, for 2.5 seconds) or constant light activation (~2.5 sec). We observed a

-p

maximal signal plateau at around 50 mW (data not shown). Based on our previous experience

re

with evoked release of glutamate this was likely due to two major factors: 1) the density of synapses proximal to the MEA and 2) the maximal amount of light dispersion in the brain, which

lP

for blue light (488nm) is estimated at no more than 1 mm 27, 28. Furthermore, the temporal dynamics of glutamate release produced by ChR2 activation were similar to what we have

24, 29-32

ur na

previously reported using other forms of stimulation (high potassium, drug induced or behavior) . The dose-response of the glutamate signals vs. light power seen from light activation had

a wider dynamic range and precision than we have seen from other methods involving micropipette drug applications, electrical stimulation or behavioral activation 26, 33, 34. These

Jo

results support the hypothesis that the ChR2-control of glutamate release was robust and precisely regulated by the light stimulus.

3.3 Electrophysiological outcomes of ChR2 and NpHR activity in dentate gyrus granule cells (DGCs): Transfection of ChR2 in rat dentate gyrus resulted in prominent labeling of principal,

12

presumably glutamatergic neurons as well as presumptive GABAergic interneurons. Transfection of NpHR in the rat dentate gyrus resulted in prominent co-immunolabeling with GABAergic neurons (Figure 2B, C); hilar mossy cells were also likely transfected. The functional outcomes of optical stimulation are important to our understanding of how these neurons affect the dentate gyrus circuit. For this study, we used optical stimulation while recording from DGCs to assess the functional outcomes of transfection of these channels into the

of

dentate gyrus of the hippocampus. Stimulation in slices from rats transfected with AAV2/5-Syn-

ro

ChR2-EYFP resulted in direct inward currents in the recorded DGCs, which exhibited an

increase in charge transfer during 16 msec to 500 msec stimulation, with no stimulus time-

-p

related differences in peak amplitude (peak amplitude 16ms stim= 143.73± 46.99 pA, peak

re

amplitude 50ms stim= 160.62± 55.54 pA, peak amplitude 500ms stim= 152.37± 50.70 pA; oneway ANOVA, F(2,27)= 0.03, p=0.97. In contrast, since responses were sustained during the light

lP

stimulus, the electrical charge over time (i.e., the charge transfer) dramatically increased as pulse width increased. This increase in charge transfer reached statistical significance during a

ur na

comparison of the 16 msec and 500 msec pulse widths (charge transfer 16 msec stim= 1.12 ±0.71 pC/pF, charge transfer 50 msec stim= 2.00 ±0.82 pC/pF, charge transfer 500 msec stim= 9.35 ±3.67 pC/pF; one-way ANOVA, F(2,27)= 4.18, p=0.03; Tukey’s post-hoc: p=0.96, 16 msec stim vs 50 msec stim, p=0.04, 16 msec stim vs 500 msec stim, p=0.07 50 msec stim vs 500 msec stim;

Jo

n=10 cells; Figure 4A-C). Together, the relatively short latency to response, similarity of peak amplitudes, and increased charge transfer with longer pulses suggest a direct activation of the recorded DGC, as opposed to a synaptic response (response latency= 17.17± 1.28 msec; Figure 4D). These findings prompted further assessment of direct versus synaptic responses by

13

examining the sensitivity of the response to pharmacological blockade of synaptic receptors or sodium channels that are required for action potential-dependent neurotransmitter release. The large inward currents observed during activation of ChR2 were insensitive to both kynurenic acid and TTX in recorded DGCs (peak amplitude 16ms stim= 96.82± 64.21 pA, peak amplitude 50ms stim= 95.86± 62.23 pA, peak amplitude 500ms stim= 96.79± 61.62 pA; charge transfer 16 msec stim= 2.43 ±1.76 pC/pF, charge transfer 50 msec stim= 2.41 ±1.70 pC/pF, charge transfer

of

500 msec stim= 8.70 ±5.97 pC/pF; Two-way ANOVA, F(2,12)= 0.2821, p= 0.76; n=5 cells;

ro

Figure 4E-G), suggesting these currents were generated directly within the recorded granule cell, consistent with ChR2 expression in transfected DGCs. Small, transient inward currents

-p

(superimposed on the initial portion of the evoked inward currents) were sensitive to kynurenic

re

acid or TTX, consistent with a separate glutamate receptor-mediated synaptic component in some neurons.

lP

A subset of DGCs in ChR2 transfected animals were recorded while voltage-clamped at 0mV to isolate putative GABAA receptor-mediated inhibitory responses. Optical stimulation of

ur na

ChR2 resulted in transient, large outward currents with no significant difference in peak amplitude between stimulation durations (peak amplitude: 16 msec stim= 244.63 ± 109.29 pA; 50 msec stim= 246.73 ± 110.03 pA; 500 msec= 253.70± 112.08 pA; F(2,12)=0.001, p=0.99; n=5 cells; Figure 5A, B). Peak amplitude of the outward current was therefore not directly related to

Jo

stimulus duration. Unlike the inward currents evoked at -65mV, the increase in light stimulation pulse width did not result in increased outward current charge transfer in recorded DGCs (charge transfer 16 msec stim= 5.04 ±2.81 pC/pF; 50 msec stim= 5.47 ±2.81 pC/pF; 500 msec stim= 6.33 ±3.61 pC/pF; one-way ANOVA, F(2,12)= 0.04, p= 0.96). The response latency was longer than for excitatory events evoked by optical stimulation of ChR2 (response latency= 25.83± 4.50

14

msec; unpaired T-test (13)=2.482, p=0.02; Figure 5D). Evoked outward currents were sensitive to bicuculline (Figure 5E), indicating they were mediated by GABAA receptor activation. This suggests that ChR2 transfection also occurred in inhibitory neurons that project to DGCs, and/or in hilar mossy cells that mediate feedforward, di-synaptic inhibition of DGCs, in addition to the glutamatergic DGCs themselves. Optical activation of AAV2/5-Syn-NpHR-EYFP, however, resulted in GABAergic

of

inhibition of DGCs, and no direct inhibition of the DGC membrane was detected. The outward

ro

currents generated after activation of NpHR exhibited increased amplitude as stimulus duration increased from 16 to 500 msec (peak amplitude 16msec stim= 9.98± 0.91 pA; 50 msec stim=

-p

18.19± 2.98pA; 500 msec stim= 20.42± 3.13 pA; one-way ANOVA, F(2,21)= 4.64, p=0.02;

re

Tukey’s post hoc: p=0.08 16 msec stim vs 50 msec stim, p=0.02 16 msec stim vs 500 msec stim, p=0.81 50 msec stim vs 500 msec stim; n=8 cells; Figure 6A,B). Similarly, charge transfer also

lP

increased with longer stimulus pulse widths (charge transfer 16 msec stim= 0.05 ±0.01 pC/pF; 50 msec stim= 0.13 ±0.03 pC/pF; 500 msec stim= 0.18 ±0.03 pC/pF; one-way ANOVA, F(2,21)=

ur na

7.34, p= 0.004; Tukey’s post hoc: p=0.07, 16 msec stim vs 50 msec stim; p=0.003, 16 msec stim vs 500 msec stim; p=0.30, 50 msec stim vs 500 msec stim; Figure 6C). Rather than being evoked at the onset of the stimulus, outward currents evoked by activating NpHR developed immediately after the light stimulus offset. The latency from the light stimulus offset to outward

Jo

current onset was similar to that for synaptic responses evoked after activation of inhibitory neurons by ChR2 (onset latency= 24.81 ±1.36 msec, Figure 6D). This suggested they resulted from the removal of the inhibitory NpHR stimulus. The light-evoked outward currents were also sensitive to bicuculline (Figure 6E), indicating they were mediated by GABAA receptors and consistent with rebound from NpHR-mediated inhibition of local inhibitory GABA neurons that

15

projected to the recorded DGCs. These data are consistent with immunolabeling, which demonstrated a predominant NpHR transfection into GABAergic neurons (Figure 2).

3.4 Behavioral Responses to ChR2- and NpHR-optogenetic stimulation: Five rats were transfected with ChR2 injected into DG. After 6 weeks, they received twice-daily stimuli with 50 mW blue light. Two developed Racine Stage 1 seizures with seven stimuli before pulling off

of

their MEA pedestals. One rat progressed to Stage 2 after 27 stimuli and one to Stage 3 after 15

ro

stimuli. Finally, one rat had two consecutive Stage 5 seizures with the 18th and 19th stimuli.

Stimulus-induced release of neurotransmitter from synaptic vesicles is facilitated by docking to

-p

and fusion of the vesicle membrane with the presynaptic plasma membrane, mediated by integral

re

membrane proteins called SNAREs. In neurons, the 7S SNARE complex (7SC) is minimally required for vesicle/plasma membrane fusion 35; formation of stable 7SC is believed to represent

lP

one of the last steps before membrane fusion and is thus a hallmark of synaptic vesicles that are in a “ready-release” state. The 7SC hippocampal ipsilateral/contralateral ratio of the rat that

ur na

experienced two consecutive Stage 5 seizures was 1.28, consistent with enhanced stimulusinduced synaptic transmission and consistent with our studies of fully electrically kindled rats stimulated from diverse sites 12, 18-21.

We also evaluated the effect of yellow light stimulation of NpHR to supress

Jo

epileptogenesis in the kindling model (Figure 7). Nine rats were DG-transfected with NpHR. After 5 weeks, rats were implanted with kindling electrodes into right basal amygdala and a guide cannula for the fiber optic cable into right DG. One week later, rats (n=5) were then stimulated twice/day: yellow light was applied for 2-3 sec followed immediately by a 1 sec kindling stimulation at the predetermined after-discharge threshold, such that laser stimulation

16

temporally overlapped kindling stimulation. Controls (n=4) received only the 1 sec kindling stimulation twice/day. Optogenetically inhibited rats required more stimuli to achieve stage 4/5 seizures than controls (p < 0.03) or historical controls from our laboratory 12, 18, 20 and the literature 36. 4. Discussion We aimed to determine how optogenetically-mediated neural network changes promote

of

or inhibit hippocampal epileptic network activity and epileptogenesis. Although this study did

ro

not aim to determine specific component mechanisms at hierarchies below the network level, results provide important information on how the evolution of a pathologic network can be

-p

enhanced or impeded by altering neuronal activity. Further, studies directed at the hierarchical

re

level of network interactions may augment interpretation of outcomes from reductionist studies that are typically used to identify specific anatomic, molecular, or genetic mechanisms. Notably,

lP

hierarchical relationships are not necessarily linear and therefore prediction of network or system behavior as a function of changes in cell-type specific activity is inherently uncertain, and

ur na

understanding epileptogenic network activity is therefore informative for understanding effects of cell-type specific pathologies. It is also important in the context of developing translationally relevant therapies for complex, multi-hierarchical disorders like epilepsy that involve cellular, network, and system pathologies.

Jo

Serotype-specific adeno-associated viruses (AAV) can be used to transfect cells in vivo

via stereotaxic injection. Selective neurotropism of specific AAV serotypes can help delimit transfection and choice of promoters can restrict expression of the opsin protein. Thus, this technique, utilizing a highly focal light stimulus to limit the area of activated opsins, allows for considerable cell type specificity with high temporal and spatial precision. The extent of viral

17

spread and cell tropism can be quite different among serotypes, dependent on target tissue and species. We used a recombinant AAV2/5 serotype, that is, the vectors contain AAV2 terminal repeats flanking the opsin-recorder cassette under the control of the synuclein promoter in an AAV5 capsid. This serotype has been shown to transfect almost the entire rostral to caudal extent of the dorsal hippocampal formation, including the DG granule cell layer and neurons within the entire pyramidal layer in the CA1-CA3 region 37, consistent with our observations (Figure 2).

of

The AAV5 capsid, which binds sialic acid on the cell surface and uses platelet-derived growth

ro

factor (PDGFR) for entry 38, 39 , restricts recorder (GFP) expression to neurons when used with the SynI neuronal synapsin promoter. 40, 41 Data presented here indicate that AAV serotype 2/5 at

-p

the titer range we used transfects excitatory and inhibitory neurons in the hippocampal

re

formation.

Several promoters have been intensively studied. 37, 42, 43 α-Calcium/calmodulin-

lP

dependent kinase II (αCamKII) and SynI are most commonly used for neuronal transfection. Both promoters differentially transfect multiple neuronal types: GFP reporter is seen in both

ur na

parvalbumin-positive GABA neurons and in large excitatory pyramidal cells independent of several AAV serotypes and using either neuronal promoter, but the expression levels of the transduced neurons is variable 43. Interestingly, Nathanson et al. reported that both SynI and αCamKII promoters in an AAV1 serotype demonstrated an excitatory/inhibitory preference that

Jo

changed by titer of the viral solution. 44 Differential levels of expression in at least two neuronal types may explain the electrophysiological data we present here. In this regard, it is well recognized that relatively small numbers of hippocampal GABA neurons, as demonstrated in Figure 2B and C, can drive an excitatory synchronous volley and inhibiting restricted numbers of GABA neurons can increase evoked seizure threshold and shorten seizure duration 6, 45, 46.

18

The apparent spread of virus we observed (Figure 2) helps ensure a wide regional, if not homogenous, level of expression among neurons. The cell-preferring but not cell-specific neurotropism of our AAV2/5 serotype helps delimit transfection to neurons, and our choice of promoter restricts expression of the opsin protein but does not alter constitutive cellular function. Because of the apparent (and fortuitous) differential expression of these channels among cell types, this model system with a single transfection vector is uniquely positioned to amplify and

of

perturb both excitatory and inhibitory circuits that participate in network changes associated with

ro

acquired epileptogenesis. These characteristics can be exploited to optogenetically induce (gainof-function) or inhibit (loss-of-function) epileptogenesis and relate these effects to network

-p

change, and to specific behavioral, cellular, electrophysiological and molecular outcomes. This

re

system is especially useful in isolating the role of network activity in epileptogenesis, as only those cells expressing the particular opsin exposed to the correct wavelength of light will

lP

respond. Other factors like cell death of vulnerable neuron populations or robust mossy fiber sprouting that are observed in animal models of epilepsy are less likely to occur in this system 47, .

ur na

48

Electrical kindling paradigms using amygdala, perforant path, or hippocampus as the stimulation site all require considerably fewer stimuli to reach full kindling than we observed using our optogenetic technique 12. Stimulation sites external to and direct electrical stimulation

Jo

of the dentate gyrus result in a large synchronous depolarization volley arising there and a presumed more rapid conversion to pathological epileptiform networks. We postulate that use of our technique results in a smaller initial region of synchronization, a longer interval to recruit additional neuronal populations, and a slower development of necessary network changes as observed in our behavioral study. Such slowed development is reminiscent of focal

19

epileptogenesis that develops after stroke and other focal injuries 49, 50. Optogenetic kindling may allow for more controlled study of cellular, electrophysiological, and network changes of epileptogenesis, analogous to optogenetics employed in studies of seizure initiation and maintenance6, 51. Interestingly, not only did the transfection of the AAV2/5 viral vectors used in this study differ in neuron populations that were transfected, but also the functional outcome of these

of

opsins was also distinct at the cellular and network levels. ChR2 transfection led predominantly

ro

to direct activation of the recorded DGCs that was largely unresponsive to kynurenic acid or

TTX application; small, transient evoked inward currents superimposed on the principal response

-p

were sensitive to kynurenic acid and were therefore consistent with activation of excitatory

re

synaptic terminals. This function is consistent with the ChR2 transfection of glutamatergic principal neurons and glutamatergic synaptic terminals on the recorded neurons. ChR2

lP

transfection of local, GABAergic neurons was also evident, and bicuculline-sensitive IPSCs were evoked in DGCs, an effect that could be mediated by directly activating inhibitory

ur na

interneurons or via a feedforward/feedback microcircuit. In contrast, the transfection of NpHR primarily co-labeled with GABAergic neurons, and the activation of this opsin resulted in a transient outward current at the conclusion of the light stimulus, which was blocked by addition of the GABAA receptor antagonist, bicuculline. The blockade of this response in the presence of

Jo

bicuculline and latency of response after conclusion of the stimulus are both consistent with the targeting of this viral transfection to GABAergic neurons in the dentate gyrus. Responses were not tested in other principal cells of the hippocampus (e.g., CA1 or CA3 pyramidal cells), and it remains possible that other hippocampal cell types will express the opsin and contribute to the inhibitory effect on kindling of NpHR activation. Interestingly, in optogenetically-evoked

20

neocortical seizures, inhibitory neurons are recruited prior to excitatory neurons at seizure onset, and optogenetic inhibition of GABA neuron subtypes can inhibit seizure initiation or duration, possibly via disinhibition of local GABA circuitry6. Further studies to identify transfection prevalence in subpopulations of hippocampal GABA neurons may be beneficial in the use of this opsin to study the process of acquired epileptogenesis, including after TBI, stroke, and CNS infection. The demonstrated feasibility of optogenetic stimulation to alter neuronal circuit and

of

system function in vivo and ex vivo shows promise for modifying epileptogenesis, but specific

ro

neural circuit interactions should also be assessed in the context of these behavioral and neural network outcomes as studies and trials move forward.

-p

To date, little work has been done using optogenetic techniques to study epileptogenesis

re

and the associated network changes. Previous work in optogenetic activation of brain regions has demonstrated the ability to evoke or suppress seizures or seizure-like activity52-55. However, this

lP

study is the first to use in vivo real time measurement of glutamate with MEAs to assess the outcome of optical stimulation in transfected rodents 56. Detection of glutamate release that is

ur na

sensitive to both stimulation frequency and duration demonstrates the ability of these opsins to respond to variable stimuli and potentials as an adaptable response, which in these studies was epileptogenesis or its hindrance.

In a recent review, Chow and Boyden propose that optogenetics be used as a therapeutic

Jo

intervention in conditions for which a clear benefit may be obtained from switching on or off specific cell types, and for which there is no better alternative. 57 Examples included retinitis pigmentosa, epilepsy, and spinal cord injury. Optogenetic therapeutics currently require the delivery of genetic constructs by means of a viral vector and once inside the body, bacterial or algal opsins may need to be stably expressed for weeks, months or even years, and this could

21

trigger an adaptive glial or immune response. That said gene therapy for Parkinson’s disease using AAV vectors has been advancing for the last decade, with continued refinements as knowledge advances. A recent clinical trial reported no clinically meaningful adverse events attributable to intraputaminal AAV2-neurturin, indwelling for up to two years. 58 Instrumentation such as Deep Brain Stimulation electrodes for Parkinson’s disease, Essential Tremor and dystonias (FDA-approved since 1999) and Vagal Nerve Stimulation for epilepsy (FDA-approved

of

in 2005) can be safely implanted and function for years. Preclinical studies using optogenetic

ro

approaches to develop new therapeutics to affect brain disorders are in process.

Optogenetic stimulation to drive behavioral alteration has been successfully implemented

-p

using in vivo electrophysiological monitoring methods. Using opsin-transfected hippocampal

re

cells, Krook-Magnuson et al. have used a closed-loop system to record real-time epileptiform activity in hippocampus and stop spontaneous temporal lobe seizures by light stimulation.53 A

lP

similar closed-loop paradigm has been used by Paz et al. to optogenetically control thalamus and interrupt seizures after cortical injury.54 However, to our knowledge few studies have been

ur na

reported that connect optogenetic stimulation to neuronal activation by monitoring neurotransmitter concentrations. Microdialysis allows direct measurements of neurotransmitter output in response to optogenetic stimulation, but does not provide the necessary high spatial and temporal resolution required to study the rapid phasic activity of glutamate (or other

Jo

neurotransmitter) release.59 In vivo amperometry using MEAs, however, does allow for quantification of extracellular neurotransmitter levels at the temporal and spatial resolution of electrophysiologic recordings. As shown here and elsewhere, this method allows for real-time measurement of both phasic and tonic glutamate release.18, 24, 60, 61 A real-time electrochemical recording device has been used to study optogenetically controlled dopamine release in PC12

22

cell cultures 62 and striatum of anesthetized rat 63, and acetylcholine in rat basal forebrain.64 The measurements of glutamate using optogenetics coupled to the MEA technology in this study are an extension of the recently reported studies by Viereckel and colleagues in the mouse globus pallidus.65 Viereckel and coworkers used viral injection into the subthalamic nucleus to produce a selective expression of channelrhodopsin via glutamate fibers to the globus pallidus. Light activation of the globus pallidus adjacent to the MEA produced rapid and transient overflow of

of

glutamate that was light intensity-dependent.65 In the present studies, we used a different viral

ro

construct that targeted neurons into the dentate gyrus of the rat hippocampus to produce

channelrhodopsin expression in the hippocampal trisynaptic pathway. Optical stimulation of

-p

ChR2 produced transient release of glutamate somewhat analogous to the work of Viereckel and

re

coworkers. However, there were two major differences observed not withstanding we used a different viral construct with a synapsin promotor and a different viral serotype. First, our light-

lP

evoked signals were very reproducible and were different in signal shape. Secondly, in comparison to Viereckel et al., our laser light pulses did not produce any photovoltaic effect or

ur na

artifacts when the recording assembly was tested in vitro and we observed visible artifacts in the hippocampus of treatment naïve rats only at light intensities < 10 mW. The artifact/photovoltaic effects were different in shape and time course from those described by Viereckel and coworkers and were easily removed using the self-referencing sentinel sites. Thus, no correction was

Jo

needed for calculation of the glutamate concentrations as seen in Viereckel et al.65 Some of these differences are likely due to the different viral constructs employed in the two different studies, the orientation of the MEA to the optical fiber and the different brain regions studied. 5. Conclusions This study demonstrates the integration of transfected opsin channels into a rodent model

23

with both in vivo and ex vivo functional outcome measures. Coupled with MEA real-time monitoring, these findings will provide a foundation for better understanding of how neuron population activity selectively contributes to disease progression and upon which future use of optogenetic interventions to modify neurological diseases, including epilepsy, can be based. Treatments affecting individual components of the disorder (e.g., GABA receptor modulators, sodium channel blockers) have failed to prevent epileptogenesis, even though clearly they can be

of

effective against seizures. This proof of principle study indicates that modifying activity at the

ro

network level can modify aspects of the epileptogenic process. These findings may provide valuable insight for novel therapy development, and will inform future, more reductionist

-p

approaches aimed at functionally relevant aspects of the network-driven disease modification.

re

Financial Interests: GAG is the Owner of Quanteon, LLC that manufactures and sells FAST MK III recording system used for measuring glutamate.

1.

ur na

References

lP

Acknowledgements Supported by Veterans Administration Medical Center (SWW, JTS); DARPA N66001-09-C2080 (GAG); NIH NS088608 (BNS)

Klein P, Dingledine R, Aronica E, et al. Commonalities in epileptogenic processes from

different acute brain insults: Do they translate? Epilepsia 2018;59:37-66. Boyden ES, Zhang F, Bamberg E, Nagel G, Deisseroth K. Millisecond-timescale,

Jo

2.

genetically targeted optical control of neural activity. Nat Neurosci 2005;8:1263-1268. 3.

Deisseroth K, Etkin A, Malenka RC. Optogenetics and the circuit dynamics of

psychiatric disease. JAMA 2015;313:2019-2020.

24

4.

Govorunova EG, Sineshchekov OA, Janz R, Liu X, Spudich JL. NEUROSCIENCE.

Natural light-gated anion channels: A family of microbial rhodopsins for advanced optogenetics. Science 2015;349:647-650. 5.

Nagel G, Szellas T, Huhn W, et al. Channelrhodopsin-2, a directly light-gated cation-

selective membrane channel. Proc Natl Acad Sci U S A 2003;100:13940-13945. 6.

Khoshkhoo S, Vogt D, Sohal VS. Dynamic, Cell-Type-Specific Roles for GABAergic

Ulusoy A, Sahin G, Bjorklund T, Aebischer P, Kirik D. Dose optimization for long-term

ro

7.

of

Interneurons in a Mouse Model of Optogenetically Inducible Seizures. Neuron 2017;93:291-298.

rAAV-mediated RNA interference in the nigrostriatal projection neurons. Mol Ther

Paxinos G, Watson C. The Rat Brain in Stereotaxic Coordinates: Fifth Edition. San

Diego: Academic Press, Inc., 2005.

Zhang F, Gradinaru V, Adamantidis AR, et al. Optogenetic interrogation of neural

lP

9.

re

8.

-p

2009;17:1574-1584.

circuits: technology for probing mammalian brain structures. Nat Protoc 2010;5:439-456. Burmeister JJ, Davis VA, Quintero JE, Pomerleau F, Huettl P, Gerhardt GA.

ur na

10.

Glutaraldehyde cross-linked glutamate oxidase coated microelectrode arrays: selectivity and resting levels of glutamate in the CNS. ACS Chem Neurosci 2013;4:721-728. 11.

Burmeister JJ, Gerhardt GA. Self-referencing ceramic-based multisite microelectrodes

Jo

for the detection and elimination of interferences from the measurement of L-glutamate and other analytes. Anal Chem 2001;73:1037-1042. 12.

Matveeva EA, Vanaman TC, Whiteheart SW, Slevin JT. Asymmetric accumulation of

hippocampal 7S SNARE complexes occurs regardless of kindling paradigm. Epilepsy Res 2007;73:266-274.

25

13.

Racine RJ. Modification of seizure activity by electrical stimulation. II. Motor seizure.

Electroencephalogr Clin Neurophysiol 1972;32:281-294. 14.

Boychuk JA, Butler CR, Halmos KC, Smith BN. Enduring changes in tonic GABAA

receptor signaling in dentate granule cells after controlled cortical impact brain injury in mice. Exp Neurol 2016;277:178-189. 15.

Butler CR, Boychuk JA, Smith BN. Effects of Rapamycin Treatment on Neurogenesis

of

and Synaptic Reorganization in the Dentate Gyrus after Controlled Cortical Impact Injury in

16.

ro

Mice. Front Syst Neurosci 2015;9:163.

Butler CR, Boychuk JA, Smith BN. Differential effects of rapamycin treatment on tonic

-p

and phasic GABAergic inhibition in dentate granule cells after focal brain injury in mice. Exp

17.

re

Neurol 2016;280:30-40.

Butler CR, Boychuk JA, Smith BN. Brain Injury-Induced Synaptic Reorganization in

18.

lP

Hilar Inhibitory Neurons Is Differentially Suppressed by Rapamycin. eNeuro 2017;4. Matveeva EA, Davis VA, Whiteheart SW, Vanaman TC, Gerhardt GA, Slevin JT.

ur na

Kindling-induced asymmetric accumulation of hippocampal 7S SNARE complexes correlates with enhanced glutamate release. Epilepsia 2012;53:157-167. 19.

Matveeva EA, Price DA, Whiteheart SW, Vanaman TC, Gerhardt GA, Slevin JT.

Reduction of vesicle-associated membrane protein 2 expression leads to a kindling-resistant

Jo

phenotype in a murine model of epilepsy. Neuroscience 2012;202:77-86. 20.

Matveeva EA, Vanaman TC, Whiteheart SW, Slevin JT. Levetiracetam prevents

kindling-induced asymmetric accumulation of hippocampal 7S SNARE complexes. Epilepsia 2008;49:1749-1758.

26

21.

Matveeva EA, Whiteheart SW, Slevin JT. Accumulation of 7S SNARE complexes in

hippocampal synaptosomes from chronically kindled rats. J Neurochem 2003;84:621-624. 22.

Ledo A, Lourenco CF, Laranjinha J, Brett CM, Gerhardt GA, Barbosa RM. Ceramic-

Based Multisite Platinum Microelectrode Arrays: Morphological Characteristics and Electrochemical Performance for Extracellular Oxygen Measurements in Brain Tissue. Anal Chem 2017;89:1674-1683. Stephens ML, Quintero JE, Pomerleau F, Huettl P, Gerhardt GA. Age-related changes in

of

23.

ro

glutamate release in the CA3 and dentate gyrus of the rat hippocampus. Neurobiol Aging 2011;32:811-820.

Stephens ML, Williamson A, Deel ME, et al. Tonic glutamate in CA1 of aging rats

-p

24.

re

correlates with phasic glutamate dysregulation during seizure. Epilepsia 2014;55:1817-1825. Danbolt NC. Glutamate uptake. Prog Neurobiol 2001;65:1-105.

26.

Burmeister JJ, Pomerleau F, Palmer M, Day BK, Huettl P, Gerhardt GA. Improved

lP

25.

ceramic-based multisite microelectrode for rapid measurements of L-glutamate in the CNS. J

27.

ur na

Neurosci Methods 2002;119:163-171.

Bernstein JG, Han X, Henninger MA, et al. Prosthetic systems for therapeutic optical

activation and silencing of genetically-targeted neurons. Proc SPIE Int Soc Opt Eng 2008;6854:68540H.

Yizhar O, Fenno LE, Davidson TJ, Mogri M, Deisseroth K. Optogenetics in neural

Jo

28.

systems. Neuron 2011;71:9-34. 29.

Hunsberger HC, Rudy CC, Batten SR, Gerhardt GA, Reed MN. P301L tau expression

affects glutamate release and clearance in the hippocampal trisynaptic pathway. J Neurochem 2015;132:169-182.

27

30.

Miller EM, Quintero JE, Pomerleau F, Huettl P, Gerhardt GA, Glaser PE. Simultaneous

glutamate recordings in the frontal cortex network with multisite biomorphic microelectrodes: New tools for ADHD research. J Neurosci Methods 2015;252:75-79. 31.

Quintero JE, Day BK, Zhang Z, et al. Amperometric measures of age-related changes in

glutamate regulation in the cortex of rhesus monkeys. Exp Neurol 2007;208:238-246. 32.

Thomas TC, Hinzman JM, Gerhardt GA, Lifshitz J. Hypersensitive glutamate signaling

of

correlates with the development of late-onset behavioral morbidity in diffuse brain-injured

33.

ro

circuitry. J Neurotrauma 2012;29:187-200.

Hascup ER, Hascup KN, Stephens M, et al. Rapid microelectrode measurements and the

-p

origin and regulation of extracellular glutamate in rat prefrontal cortex. J Neurochem

34.

re

2010;115:1608-1620.

Onifer SM, Quintero JE, Gerhardt GA. Cutaneous and electrically evoked glutamate

35.

lP

signaling in the adult rat somatosensory system. J Neurosci Methods 2012;208:146-154. Weber T, Zemelman BV, McNew JA, et al. SNAREpins: minimal machinery for

36.

ur na

membrane fusion. Cell 1998;92:759-772.

Le Gal La Salle G. Amygdaloid kindling in the rat: Regional differences and general

properties. In: Wada JA, ed. Kindling 2. New York, NY: Raven Press, 1981: 31-47. 37.

Burger C, Gorbatyuk OS, Velardo MJ, et al. Recombinant AAV viral vectors

Jo

pseudotyped with viral capsids from serotypes 1, 2, and 5 display differential efficiency and cell tropism after delivery to different regions of the central nervous system. Mol Ther 2004;10:302317. 38.

Di Pasquale G, Davidson BL, Stein CS, et al. Identification of PDGFR as a receptor for

AAV-5 transduction. Nat Med 2003;9:1306-1312.

28

39.

Walters RW, Yi SM, Keshavjee S, et al. Binding of adeno-associated virus type 5 to 2,3-

linked sialic acid is required for gene transfer. J Biol Chem 2001;276:20610-20616. 40.

Kugler S, Lingor P, Scholl U, Zolotukhin S, Bahr M. Differential transgene expression in

brain cells in vivo and in vitro from AAV-2 vectors with small transcriptional control units. Virology 2003;311:89-95. 41.

Shevtsova Z, Malik JM, Michel U, Bahr M, Kugler S. Promoters and serotypes: targeting

of

of adeno-associated virus vectors for gene transfer in the rat central nervous system in vitro and

42.

ro

in vivo. Exp Physiol 2005;90:53-59.

Holehonnur R, Luong JA, Chaturvedi D, et al. Adeno-associated viral serotypes produce

-p

differing titers and differentially transduce neurons within the rat basal and lateral amygdala.

43.

re

BMC Neurosci 2014;15:28.

Watakabe A, Ohtsuka M, Kinoshita M, et al. Comparative analyses of adeno-associated

Res 2015;93:144-157.

Nathanson JL, Yanagawa Y, Obata K, Callaway EM. Preferential labeling of inhibitory

ur na

44.

lP

viral vector serotypes 1, 2, 5, 8 and 9 in marmoset, mouse and macaque cerebral cortex. Neurosci

and excitatory cortical neurons by endogenous tropism of adeno-associated virus and lentivirus vectors. Neuroscience 2009;161:441-450. 45.

Smith BN. "Maestro" Hub Neurons Orchestrate the Immature GABA Network

Jo

Symphony. Epilepsy Curr 2013;13:93-94. 46.

Yekhlef L, Breschi GL, Lagostena L, Russo G, Taverna S. Selective activation of

parvalbumin- or somatostatin-expressing interneurons triggers epileptic seizurelike activity in mouse medial entorhinal cortex. J Neurophysiol 2015;113:1616-1630.

29

47.

Shibley H, Smith BN. Pilocarpine-induced status epilepticus results in mossy fiber

sprouting and spontaneous seizures in C57BL/6 and CD-1 mice. Epilepsy Res 2002;49:109-120. 48.

Winokur RS, Kubal T, Liu D, Davis SF, Smith BN. Recurrent excitation in the dentate

gyrus of a murine model of temporal lobe epilepsy. Epilepsy Res 2004;58:93-105. 49.

Hunt RF, Boychuk JA, Smith BN. Neural circuit mechanisms of post-traumatic epilepsy.

Front Cell Neurosci 2013;7:89. Kelly KM, Kharlamov A, Hentosz TM, et al. Photothrombotic brain infarction results in

of

50.

51.

ro

seizure activity in aging Fischer 344 and Sprague Dawley rats. Epilepsy Res 2001;47:189-203. Ye H, Kaszuba S. Inhibitory or excitatory? Optogenetic interrogation of the functional

Armstrong C, Krook-Magnuson E, Oijala M, Soltesz I. Closed-loop optogenetic

re

52.

-p

roles of GABAergic interneurons in epileptogenesis. J Biomed Sci 2017;24:93.

intervention in mice. Nat Protoc 2013;8:1475-1493.

Krook-Magnuson E, Armstrong C, Oijala M, Soltesz I. On-demand optogenetic control

lP

53.

of spontaneous seizures in temporal lobe epilepsy. Nat Commun 2013;4:1376. Paz JT, Davidson TJ, Frechette ES, et al. Closed-loop optogenetic control of thalamus as

ur na

54.

a tool for interrupting seizures after cortical injury. Nat Neurosci 2013;16:64-70. 55.

Sukhotinsky I, Chan AM, Ahmed OJ, et al. Optogenetic delay of status epilepticus onset

in an in vivo rodent epilepsy model. PLoS One 2013;8:e62013. Parrot S, Denoroy L, Renaud B, Benetollo C. Why Optogenetics Needs in Vivo

Jo

56.

Neurochemistry. ACS Chem Neurosci 2015;6:948-950. 57.

Chow BY, Boyden ES. Optogenetics and translational medicine. Sci Transl Med

2013;5:177ps175.

30

58.

Olanow CW, Bartus RT, Baumann TL, et al. Gene delivery of neurturin to putamen and

substantia nigra in Parkinson disease: A double-blind, randomized, controlled trial. Ann Neurol 2015;78:248-257. 59.

Zant JC, Kim T, Prokai L, et al. Cholinergic Neurons in the Basal Forebrain Promote

Wakefulness by Actions on Neighboring Non-Cholinergic Neurons: An Opto-Dialysis Study. J Neurosci 2016;36:2057-2067. Batten SR, Matveeva EA, Whiteheart SW, Vanaman TC, Gerhardt GA, Slevin JT.

of

60.

the STXBP5/tomosyn-1 gene. Brain Behav 2017;7:e00795.

Viereckel T, Dumas S, Smith-Anttila CJ, et al. Midbrain Gene Screening Identifies a

-p

61.

ro

Linking kindling to increased glutamate release in the dentate gyrus of the hippocampus through

re

New Mesoaccumbal Glutamatergic Pathway and a Marker for Dopamine Cells Neuroprotected in Parkinson's Disease. Sci Rep 2016;6:35203.

Chiu WT, Lin CM, Tsai TC, et al. Real-time electrochemical recording of dopamine

lP

62.

release under optogenetic stimulation. PLoS One 2014;9:e89293. Bass CE, Grinevich VP, Vance ZB, Sullivan RP, Bonin KD, Budygin EA. Optogenetic

ur na

63.

control of striatal dopamine release in rats. J Neurochem 2010;114:1344-1352. 64.

Gritton HJ, Howe WM, Mallory CS, Hetrick VL, Berke JD, Sarter M. Cortical

cholinergic signaling controls the detection of cues. Proc Natl Acad Sci U S A 2016;113:E1089-

Jo

1097. 65.

Viereckel T, Konradsson-Geuken A, Wallen-Mackenzie A. Validated multi-step

approach for in vivo recording and analysis of optogenetically evoked glutamate in the mouse globus pallidus. J Neurochem 2018;145:125-138.

31

-p

ro

of

Figures

Jo

ur na

lP

re

Figure 1: MEA with attached fiber optic cannula and glass pipette.

32

of

Figure 2. Distribution of Transfected AAV Constructs. A. ChR2-YFP expression in DG

ro

(10X) showing bilateral distribution of AAV2/5-CHR2-hSyn-EYFP in the hippocampus 6 weeks

-p

after a single injection (1 μl) into the right DG. B. AAV2/5-hSyn-NpHR 3.0-EYFP transfection (EYFP, green) and GAD67 immunolabeling (red) in the DG of a different rat (10X). There is a

re

predominant colocalization of EYFP and GAD67 label, consistent with preferential transfection

Jo

ur na

labeled hilar interneuron.

lP

of GABAergic, inhibitory neurons. C. Enlargement of the boxed area in B, centered on a co-

33

of ro -p re lP

Figure 3. Light intensity dependent increases in glutamate release. Blue laser-induced

ur na

release of glutamate in rat DG using MEA + waveguide array. Comparison of pulse trains (100 pulses) with variable pulse durations vs. constant blue laser activations. A range of 0.2-50 mW laser intensities were used to produce reproducible light intensity-dependent,

Jo

increases in extracellular glutamate

34

of ro

-p

Figure 4: Channelrhodopsin (ChR2)-activated inward currents in dentate granule cells. A. Representative recording of response in dentate granule cell (DGC) held at -65mV to blue light

re

in rat transfected with ChR2. B. Bar graph of peak amplitude of responses to ChR2 activity at 3

lP

stimulation durations 16 msec, 50 msec, and 500 msec. C. Bar graph of charge transfer normalized to whole cell capacitance measures for direct ChR2 activity at 16 msec, 50 msec, and

ur na

500 msec durations. D. Bar graph of latency to peak response following blue light onset. E. Representative trace of recorded DGC response to blue light in the presence of kynurenic acid and tetrodotoxin in circulating ACSF. F. Bar graph of peak amplitude responses of DGCs in nACSF and ACSF with kynurenic acid and tetrodotoxin added. G. Bar graph of charge transfer

Jo

normalized to whole cell capacitance in DGCs recorded in nACSF and ACSF with kynurenic acid and tetrodotoxin added. Data represented as mean ±SEM. * p<0.05

35

of

Figure 5: ChR2 stimulation elicited GABAA receptor-mediated inhibitory synaptic responses in DGCs. A. Representative trace of response in DGC held at 0mV to blue light in rat

ro

transfected with ChR2. B. Bar graph of peak amplitude of responses to ChR2 activity at 3

-p

stimulation durations 16 msec, 50 msec, and 500 msec. C. Bar graph of charge transfer normalized to whole cell capacitance measures for direct ChR2 activity at 16 msec, 50 msec, and

re

500 msec durations. D. Bar graph of latency to peak response following blue light onset. E.

Jo

ur na

represented as mean ±SEM.

lP

Representative trace demonstrating sensitivity of responses to bicuculline (30 µM). Data

36

of

Figure 6: DGC responses to activation of halorhodopsin (NpHR). A. Representative trace of response in DGC held at -65mV to blue light activity in rat injected with NpHR. Outward

ro

currents developed at the offset of the inhibitory stimulus. B. Bar graph of peak amplitude of responses to NpHR activity at 3 stimulation durations 16 msec, 50 msec, and 500 msec. C. Bar

-p

graph of charge transfer normalized to whole cell capacitance measures for direct ChR2 activity

re

at 16 msec, 50 msec, and 500 msec durations. D. Bar graph of latency to peak response following blue light offset. E. Representative trace demonstrating sensitivity of responses to

Jo

ur na

lP

bicuculline. Data represented as mean ±SEM. * p<0.05

37

of ro -p

Figure 7. Mitigation of epileptogenesis by expression and activation of halorhodopsin

re

(NpHR). NpHR animals receiving overlapping yellow light activation (2-3s) during 1-sec electrical stimulation of amygdala (yellow bar) required more stimuli (P<0.03) to achieve stage

lP

4/5 seizures (21.4 ± 5.1 [SD]; n=5) than those without light activation (green bar; 11.0 ± 5.4;

Jo

ur na

n=4) or historical non-transfected controls 12, 18, 20, 21 (brown bar; 8.2 ± 3.1).

38